1
|
Lu S, Li Y, Wang L, Xiong K, Yan J, Zhai Z, Yan W. Effects of Herpud1 in Methamphetamine-induced Neuronal Apoptosis. Curr Med Chem 2025; 32:1406-1422. [PMID: 38299291 DOI: 10.2174/0109298673277857231221110453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Methamphetamine (METH) is an illicit psychoactive substance that can damage various organs in the body, especially the nervous system. We hypothesized that expression of homocysteine-inducible endoplasmic reticulum-resident with ubiquitin-like domain member 1 (Herpud1) protein would alleviate the induction of apoptosis following METH administration. METHODS To test this hypothesis, we analysed the changes in Herpud1 expression and apoptosis in PC12 cells under different concentrations and exposure times of METH. Moreover, we examined the effects of Herpud1 knockdown on METH-induced neuronal apoptosis. Flow cytometry and Western blot analyses were used to evaluate apoptosis levels and the expression of apoptotic markers (cleaved caspase-3) in PC12 cells following Herpud1 knockdown by synthetic small interfering RNA (siRNA). RESULTS Our results showed that Herpud1 expression was upregulated in PC12 cells following METH treatment, while endoplasmic reticulum stress (ERS) and apoptosis were also increased. Conversely, Herpud1 knockdown reduced METH-induced ERS and apoptosis levels in vitro. CONCLUSION These results suggest that Herpud1 plays an essential role in METH-induced neuronal ERS and apoptosis and may represent a potential therapeutic gene target in METH-induced neurotoxicity.
Collapse
Affiliation(s)
- Shuang Lu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Yan Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Lewen Wang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Zhihao Zhai
- Department of Neurosurgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangzhou, 518000, China
| | - Weitao Yan
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
2
|
Xia J, Wang J, Zhao N, Zhang Q, Xu B. Effects of treadmill exercise on endoplasmic reticulum protein folding and endoplasmic reticulum-associated protein degradation pathways in APP/PS1 mice. Heliyon 2024; 10:e38458. [PMID: 39397952 PMCID: PMC11467616 DOI: 10.1016/j.heliyon.2024.e38458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
A hallmark of Alzheimer's disease (AD) is the disruption of protein homeostasis (proteostasis), manifested by the misfolding and aggregation of proteins. Molecular chaperones and the endoplasmic reticulum (ER)-associated protein degradation (ERAD) pathway in the ER are essential for correct protein folding and degradation of misfolded proteins respectively, thus contributing to the maintenance of proteostasis. The present study aimed to investigate whether the beneficial effects of exercise in an AD mice model is associated with changes in ER protein folding and ERAD. APP/PS1 transgenic and wild-type mice were subjected to treadmill exercise for three months. The levels of molecular chaperones, specifically protein disulfide isomerases (PDIs) and heat shock proteins (HSPs), as well as ERAD-associated molecules were analyzed in the hippocampus. The result revealed a decrease in mRNA levels of PDIA2, PDIA3, PDIA4, PDIA5, PDIA6, HSPA1B, HSPA8, HSP90B1, DNAJB2, CRYAB, and CNX, an increase in mRNA levels of HSPA5 and HSPH1, an increase in protein levels of HERPUD1, and a decrease in protein levels of VCP in APP/PS1 mice. However, following a 3-month treadmill exercise regimen, an increase in mRNA levels of PDIA2, PDIA4, PDIA6, HSPA1A, HSPA8, HSP90AB1, and DNAJB2, as well as an increase in protein levels of VCP and DERL2, and a decrease in protein levels of HERPUD1 were noted. Overall, our findings indicate that disruptions in hippocampal ER protein folding and ERAD pathways may be implicated in AD, with exercise serving as a regulator of these pathways.
Collapse
Affiliation(s)
- Jie Xia
- Department of Physical Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jing Wang
- Research Center of Basic Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Na Zhao
- College of Sports and Health, Shandong Sport University, Jinan, 250102, China
| | - Qiang Zhang
- Genetics and Genomic Medicine Research and Teaching Department, University College London, London, WC1E 6BT, United Kingdom
| | - Bo Xu
- College of Physical Education and Health, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
3
|
Lin LL, Wang HH, Pederson B, Wei X, Torres M, Lu Y, Li ZJ, Liu X, Mao H, Wang H, Zhou LE, Zhao Z, Sun S, Qi L. SEL1L-HRD1 interaction is required to form a functional HRD1 ERAD complex. Nat Commun 2024; 15:1440. [PMID: 38365914 PMCID: PMC10873344 DOI: 10.1038/s41467-024-45633-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/30/2024] [Indexed: 02/18/2024] Open
Abstract
The SEL1L-HRD1 protein complex represents the most conserved branch of endoplasmic reticulum (ER)-associated degradation (ERAD). Despite recent advances in both mouse models and humans, in vivo evidence for the importance of SEL1L in the ERAD complex formation and its (patho-)physiological relevance in mammals remains limited. Here we report that SEL1L variant p.Ser658Pro (SEL1LS658P) is a pathogenic hypomorphic mutation, causing partial embryonic lethality, developmental delay, and early-onset cerebellar ataxia in homozygous mice carrying the bi-allelic variant. Biochemical analyses reveal that SEL1LS658P variant not only reduces the protein stability of SEL1L, but attenuates the SEL1L-HRD1 interaction, likely via electrostatic repulsion between SEL1L F668 and HRD1 Y30 residues. Proteomic screens of SEL1L and HRD1 interactomes reveal that SEL1L-HRD1 interaction is a prerequisite for the formation of a functional HRD1 ERAD complex, as SEL1L is required for the recruitment of E2 enzyme UBE2J1 as well as DERLIN to HRD1. These data not only establish the disease relevance of SEL1L-HRD1 ERAD, but also provide additional insight into the formation of a functional HRD1 ERAD complex.
Collapse
Affiliation(s)
- Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Huilun Helen Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Brent Pederson
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Mauricio Torres
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - You Lu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Xiaodan Liu
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Hancheng Mao
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Hui Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Linyao Elina Zhou
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Shengyi Sun
- Department of Pharmacology, University of Virginia, School of Medicine, Charlottesville, VA, 22908, USA.
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
4
|
Christianson JC, Jarosch E, Sommer T. Mechanisms of substrate processing during ER-associated protein degradation. Nat Rev Mol Cell Biol 2023; 24:777-796. [PMID: 37528230 DOI: 10.1038/s41580-023-00633-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 08/03/2023]
Abstract
Maintaining proteome integrity is essential for long-term viability of all organisms and is overseen by intrinsic quality control mechanisms. The secretory pathway of eukaryotes poses a challenge for such quality assurance as proteins destined for secretion enter the endoplasmic reticulum (ER) and become spatially segregated from the cytosolic machinery responsible for disposal of aberrant (misfolded or otherwise damaged) or superfluous polypeptides. The elegant solution provided by evolution is ER-membrane-bound ubiquitylation machinery that recognizes misfolded or surplus proteins or by-products of protein biosynthesis in the ER and delivers them to 26S proteasomes for degradation. ER-associated protein degradation (ERAD) collectively describes this specialized arm of protein quality control via the ubiquitin-proteasome system. But, instead of providing a single strategy to remove defective or unwanted proteins, ERAD represents a collection of independent processes that exhibit distinct yet overlapping selectivity for a wide range of substrates. Not surprisingly, ER-membrane-embedded ubiquitin ligases (ER-E3s) act as central hubs for each of these separate ERAD disposal routes. In these processes, ER-E3s cooperate with a plethora of specialized factors, coordinating recognition, transport and ubiquitylation of undesirable secretory, membrane and cytoplasmic proteins. In this Review, we focus on substrate processing during ERAD, highlighting common threads as well as differences between the many routes via ERAD.
Collapse
Affiliation(s)
- John C Christianson
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| | - Ernst Jarosch
- Max-Delbrück-Centrer for Molecular Medicine in Helmholtz Association, Berlin-Buch, Germany
| | - Thomas Sommer
- Max-Delbrück-Centrer for Molecular Medicine in Helmholtz Association, Berlin-Buch, Germany.
- Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
5
|
Son SH, Lee J, Cho SN, Choi JA, Kim J, Nguyen TD, Lee SA, Son D, Song CH. Herp regulates intracellular survival of Mycobacterium tuberculosis H37Ra in macrophages by regulating reactive oxygen species-mediated autophagy. mBio 2023; 14:e0153523. [PMID: 37800958 PMCID: PMC10653826 DOI: 10.1128/mbio.01535-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/21/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE Several studies have suggested that endoplasmic reticulum (ER) stress is important in the pathogenesis of infectious diseases; however, the precise function of ER stress regulation and the role of Herp as a regulator in Mtb H37Ra-induced ER stress remain elusive. Therefore, our study investigated ER stress and autophagy associated with Herp expression in Mycobacterium tuberculosis-infected macrophages to determine the role of Herp in the pathogenesis of tuberculosis.
Collapse
Affiliation(s)
- Sang-Hun Son
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Junghwan Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, South Korea
| | - Soo-Na Cho
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Ji-Ae Choi
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, South Korea
| | - Jaewhan Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Tam Doan Nguyen
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seong-Ahn Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Doyi Son
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chang-Hwa Song
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
- Translational Immunology Institute, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
6
|
Cremer T, Voortman LM, Bos E, Jongsma MLM, ter Haar LR, Akkermans JJLL, Talavera Ormeño CMP, Wijdeven RHM, de Vries J, Kim RQ, Janssen GMC, van Veelen PA, Koning RI, Neefjes J, Berlin I. RNF26 binds perinuclear vimentin filaments to integrate ER and endolysosomal responses to proteotoxic stress. EMBO J 2023; 42:e111252. [PMID: 37519262 PMCID: PMC10505911 DOI: 10.15252/embj.2022111252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Proteotoxic stress causes profound endoplasmic reticulum (ER) membrane remodeling into a perinuclear quality control compartment (ERQC) for the degradation of misfolded proteins. Subsequent return to homeostasis involves clearance of the ERQC by endolysosomes. However, the factors that control perinuclear ER integrity and dynamics remain unclear. Here, we identify vimentin intermediate filaments as perinuclear anchors for the ER and endolysosomes. We show that perinuclear vimentin filaments engage the ER-embedded RING finger protein 26 (RNF26) at the C-terminus of its RING domain. This restricts RNF26 to perinuclear ER subdomains and enables the corresponding spatial retention of endolysosomes through RNF26-mediated membrane contact sites (MCS). We find that both RNF26 and vimentin are required for the perinuclear coalescence of the ERQC and its juxtaposition with proteolytic compartments, which facilitates efficient recovery from ER stress via the Sec62-mediated ER-phagy pathway. Collectively, our findings reveal a scaffolding mechanism that underpins the spatiotemporal integration of organelles during cellular proteostasis.
Collapse
Affiliation(s)
- Tom Cremer
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Lenard M Voortman
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik Bos
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Marlieke LM Jongsma
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Laurens R ter Haar
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jimmy JLL Akkermans
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Cami MP Talavera Ormeño
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Ruud HM Wijdeven
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam NeuroscienceAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Jelle de Vries
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Robbert Q Kim
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - George MC Janssen
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Peter A van Veelen
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Roman I Koning
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Ilana Berlin
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
7
|
Su R, Yin J, Ruan X, Chen Y, Wan P, Luo Z. Featured interactome of homocysteine-inducible endoplasmic reticulum protein uncovers novel binding partners in response to ER stress. Comput Struct Biotechnol J 2023; 21:4478-4487. [PMID: 37736299 PMCID: PMC10510068 DOI: 10.1016/j.csbj.2023.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Homocysteine-inducible endoplasmic reticulum protein (HERP) is an endoplasmic reticulum (ER)-resident protein and important for the adaptation of cellular protein homeostasis by ER-associated degradation (ERAD) system. HERP interactors are critical for cellular viability and the reaction to ER stress. To explore the exact mechanisms by which HERP performed the biological functions, we conducted an interaction analysis of HERP protein in HeLa cells by co-immunoprecipitation (Co-IP) and liquid chromatography-mass spectrometer (LC-MS)/MS coupled with label-free quantification (LFQ). Among the interactome results, 123 proteins significantly interacted with HERP, which leads to numerous biological processes including protein import into nucleus, ubiquitin-dependent ERAD pathway, negative regulation of apoptotic process, and protein transport from ER, along with multiple pathways including several diseases, protein processing in ER, fatty acid metabolism, and steroid biosynthesis. Furthermore, we selected several prey proteins from the interactome data and confirmed that HERP interacted with ancient ubiquitous protein 1 (AUP1), Fas-associated factor family member 2 (FAF2), tripartite motif containing 47 (TRIM47), acyl-CoA synthetase long-chain family member 3 (ACSL3), sequestosome 1 (SQSTM1), and poly(rC) binding protein 2 (PCBP2) by Co-IP and confocal microscopy experiments, respectively. Moreover, the expression and location of several interacted proteins were obviously altered in response to ER stress induced by Thapsigargin stimulation and Enterovirus 71 infection. In conclusion, our findings revealed that the vital proteins interacted with HERP to mediate signaling transduction, thus providing novel clues for the mechanisms of HERP associated with ERAD and metabolism in response to ER stress under physiological and pathological conditions.
Collapse
Affiliation(s)
- Rui Su
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang 453003, China
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Jialing Yin
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Xiaolan Ruan
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Yanxi Chen
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Pin Wan
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430072, China
- Foshan Institute of Medical Microbiology, Foshan 528315, China
| | - Zhen Luo
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
- Foshan Institute of Medical Microbiology, Foshan 528315, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| |
Collapse
|
8
|
Badawi S, Mohamed FE, Varghese DS, Ali BR. Genetic disruption of mammalian endoplasmic reticulum-associated protein degradation: Human phenotypes and animal and cellular disease models. Traffic 2023; 24:312-333. [PMID: 37188482 DOI: 10.1111/tra.12902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
Endoplasmic reticulum-associated protein degradation (ERAD) is a stringent quality control mechanism through which misfolded, unassembled and some native proteins are targeted for degradation to maintain appropriate cellular and organelle homeostasis. Several in vitro and in vivo ERAD-related studies have provided mechanistic insights into ERAD pathway activation and its consequent events; however, a majority of these have investigated the effect of ERAD substrates and their consequent diseases affecting the degradation process. In this review, we present all reported human single-gene disorders caused by genetic variation in genes that encode ERAD components rather than their substrates. Additionally, after extensive literature survey, we present various genetically manipulated higher cellular and mammalian animal models that lack specific components involved in various stages of the ERAD pathway.
Collapse
Affiliation(s)
- Sally Badawi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Feda E Mohamed
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Divya Saro Varghese
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
9
|
Murase R, Yamamoto A, Hirata Y, Oh-Hashi K. Expression analysis and functional characterization of thioredoxin domain-containing protein 11. Mol Biol Rep 2022; 49:10541-10556. [PMID: 36152228 DOI: 10.1007/s11033-022-07932-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUNDS The endoplasmic reticulum (ER) is a crucial organelle that regulates both the folding, modification and transport of many proteins and senses certain stimuli inside and outside of cells. ER-associated degradation (ERAD), including SEL1L is a crucial mechanism to maintain homeostasis. In this study, we performed comparative proteome analysis in wild-type (wt) and SEL1L-deficient cells. METHODS AND RESULTS We found constitutively high expression of thioredoxin domain-containing protein 11 (TXNDC11) mRNA and protein in our SEL1L-deficient HEK293 cells by RT-PCR and Western blot analysis. The TXNDC11 gene possesses a well-conserved unfolded protein response element (UPRE) around its transcription start site, and ER stress increased TXNDC11 mRNA and luciferase reporter activity via this putative UPRE in HEK293 cells. The amounts of TXNDC11 protein in wild-type and SEL1L-deficient cells with or without thapsigargin (Tg) treatment were parallel to their mRNAs in these cells, which was almost proportional to spliced XBP1 (sXBP1) mRNA expression. The establishment and characterization of TXNDC11-deficient HEK293 cells revealed that the expression of three different ER resident stress sensors, ATF6α, CREB3 and CREB3L2, is regulated by TXNDC11. The rate of disappearance of the three proteins by CHX treatment in wt cells was remarkably different, and the full-length CREB3L2 protein was almost completely degraded within 15 min after CHX treatment. TXNDC11 deficiency increased the expression of each full-length form under resting conditions and delayed their disappearance by CHX treatment. Interestingly, the degree of increase in full-length CREB3/CREB3L2 by TXNDC11 deficiency was apparently higher than that in full-length ATF6α. The increase in these proteins by TXNDC11 deficiency was hardly correlated with the expression of each mRNA. Treatment with ER stress inducers influenced each full-length mature form, and the difference in each full-length form observed in wt and TXNDC11-deficient cells was smaller. CONCLUSION This study demonstrated that TXNDC11 is an ER stress-inducible gene regulated by the IRE1-sXBP1 pathway. In addition, TXNDC11 is involved in the regulation of ATF6α, CREB3 and CREB3L2 protein expression, although the contribution to the stability of these proteins is quite variable. Therefore, its further characterization will provide new insights for understanding protein homeostasis in ER physiology and pathology.
Collapse
Affiliation(s)
- Ryoichi Murase
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Ayumi Yamamoto
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yoko Hirata
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.,Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Kentaro Oh-Hashi
- Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan. .,Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan. .,United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
| |
Collapse
|
10
|
Sun Y, Fan Y, Wang Z, Li M, Su D, Liu Y, Liang X. S100A16 promotes acute kidney injury by activating HRD1-induced ubiquitination and degradation of GSK3β and CK1α. Cell Mol Life Sci 2022; 79:184. [PMID: 35279748 PMCID: PMC8918193 DOI: 10.1007/s00018-022-04213-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/29/2022]
Abstract
AbstractThe pathogenesis of acute kidney injury (AKI) is associated with the activation of multiple signaling pathways, including Wnt/β-catenin signaling. However, the mechanism of Wnt/β-catenin pathway activation in renal interstitial fibroblasts during AKI is unclear. S100 calcium-binding protein A16 (S100A16), a new member of calcium-binding protein S100 family, is a multi-functional signaling factor involved in various pathogenies, including tumors, glycolipid metabolism disorder, and chronic kidney disease (CKD). We investigated the potential participation of S100A16 in Wnt/β-catenin pathway activation during AKI by subjecting wild-type (WT) and S100A16 knockout (S100A16+/−) mice to the ischemia–reperfusion injury (IRI), and revealed S100A16 upregulation in this model, in which knockout of S100A16 impeded the Wnt/β-catenin signaling pathway activation and recovered the expression of downstream hepatocyte growth factor (HGF). We also found that S100A16 was highly expressed in Platelet-derived growth factor receptor beta (PDGFRβ) positive renal fibroblasts in vivo. Consistently, in rat renal interstitial fibroblasts (NRK-49F cells), both hypoxia/reoxygenation and S100A16 overexpression exacerbated fibroblasts apoptosis and inhibited HGF secretion; whereas S100A16 knockdown or Wnt/β-catenin pathway inhibitor ICG-001 reversed these changes. Mechanistically, we showed that S100A16 promoted Wnt/β-catenin signaling activation via the ubiquitylation and degradation of β-catenin complex members, glycogen synthase kinase 3β (GSK3β) and casein kinase 1α (CK1α), mediated by E3 ubiquitin ligase, the HMG-CoA reductase degradation protein 1 (HRD1). Our study identified the S100A16 as a key regulator in the activation of Wnt/β-catenin signaling pathway in AKI.
Collapse
Affiliation(s)
- Yifei Sun
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ya Fan
- Department of Pathology, Nanjing Medical University, Nanjing, 211166, China
| | - Zheng Wang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Min Li
- Department of Pathology, Nanjing Medical University, Nanjing, 211166, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, 211166, China
| | - Yun Liu
- Department of Geratology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiubin Liang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
11
|
Luizet JB, Raymond J, Lacerda TLS, Barbieux E, Kambarev S, Bonici M, Lembo F, Willemart K, Borg JP, Celli J, Gérard FCA, Muraille E, Gorvel JP, Salcedo SP. The Brucella effector BspL targets the ER-associated degradation (ERAD) pathway and delays bacterial egress from infected cells. Proc Natl Acad Sci U S A 2021; 118:e2105324118. [PMID: 34353909 PMCID: PMC8364137 DOI: 10.1073/pnas.2105324118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Perturbation of the endoplasmic reticulum (ER), a central organelle of the cell, can have critical consequences for cellular homeostasis. An elaborate surveillance system known as ER quality control ensures that cells can respond and adapt to stress via the unfolded protein response (UPR) and that only correctly assembled proteins reach their destination. Interestingly, several bacterial pathogens hijack the ER to establish an infection. However, it remains poorly understood how bacterial pathogens exploit ER quality-control functions to complete their intracellular cycle. Brucella spp. replicate extensively within an ER-derived niche, which evolves into specialized vacuoles suited for exit from infected cells. Here we present Brucella-secreted protein L (BspL), a Brucella abortus effector that interacts with Herp, a central component of the ER-associated degradation (ERAD) machinery. We found that BspL enhances ERAD at the late stages of the infection. BspL targeting of Herp and ERAD allows tight control of the kinetics of autophagic Brucella-containing vacuole formation, delaying the last step of its intracellular cycle and cell-to-cell spread. This study highlights a mechanism by which a bacterial pathogen hijacks ERAD components for fine regulation of its intracellular trafficking.
Collapse
Affiliation(s)
- Jean-Baptiste Luizet
- Laboratory of Molecular Microbiology and Structural Biochemistry, CNRS UMR5086, Université de Lyon, 69367 Lyon, France
| | - Julie Raymond
- Laboratory of Molecular Microbiology and Structural Biochemistry, CNRS UMR5086, Université de Lyon, 69367 Lyon, France
| | - Thais Lourdes Santos Lacerda
- Laboratory of Molecular Microbiology and Structural Biochemistry, CNRS UMR5086, Université de Lyon, 69367 Lyon, France
| | - Emeline Barbieux
- Department of Biology, Research Unit in Microorganisms Biology, Namur Research Institute for Life Sciences, 5000 Namur, Belgium
- Laboratory of Parasitology, Université Libre de Bruxelles Centre for Research in Immunology (UCRI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Stanimir Kambarev
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164
| | - Magali Bonici
- Laboratory of Molecular Microbiology and Structural Biochemistry, CNRS UMR5086, Université de Lyon, 69367 Lyon, France
| | - Frédérique Lembo
- Equipe labellisée Ligue 'Cell Polarity, Cell Signaling and Cancer', Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, CNRS, INSERM, 13009 Marseille, France
| | - Kévin Willemart
- Department of Biology, Research Unit in Microorganisms Biology, Namur Research Institute for Life Sciences, 5000 Namur, Belgium
| | - Jean-Paul Borg
- Equipe labellisée Ligue 'Cell Polarity, Cell Signaling and Cancer', Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Aix-Marseille Université, CNRS, INSERM, 13009 Marseille, France
- Institut Universitaire de France, 75231 Paris, France
| | - Jean Celli
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164
| | - Francine C A Gérard
- Laboratory of Molecular Microbiology and Structural Biochemistry, CNRS UMR5086, Université de Lyon, 69367 Lyon, France
| | - Eric Muraille
- Department of Biology, Research Unit in Microorganisms Biology, Namur Research Institute for Life Sciences, 5000 Namur, Belgium
- Laboratory of Parasitology, Université Libre de Bruxelles Centre for Research in Immunology (UCRI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy, CNRS, INSERM, Aix-Marseille Université, 13009 Marseille, France
| | - Suzana P Salcedo
- Laboratory of Molecular Microbiology and Structural Biochemistry, CNRS UMR5086, Université de Lyon, 69367 Lyon, France;
| |
Collapse
|
12
|
Ninagawa S, George G, Mori K. Mechanisms of productive folding and endoplasmic reticulum-associated degradation of glycoproteins and non-glycoproteins. Biochim Biophys Acta Gen Subj 2020; 1865:129812. [PMID: 33316349 DOI: 10.1016/j.bbagen.2020.129812] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND The quality of proteins destined for the secretory pathway is ensured by two distinct mechanisms in the endoplasmic reticulum (ER): productive folding of newly synthesized proteins, which is assisted by ER-localized molecular chaperones and in most cases also by disulfide bond formation and transfer of an oligosaccharide unit; and ER-associated degradation (ERAD), in which proteins unfolded or misfolded in the ER are recognized and processed for delivery to the ER membrane complex, retrotranslocated through the complex with simultaneous ubiquitination, extracted by AAA-ATPase to the cytosol, and finally degraded by the proteasome. SCOPE OF REVIEW We describe the mechanisms of productive folding and ERAD, with particular attention to glycoproteins versus non-glycoproteins, and to yeast versus mammalian systems. MAJOR CONCLUSION Molecular mechanisms of the productive folding of glycoproteins and non-glycoproteins mediated by molecular chaperones and protein disulfide isomerases are well conserved from yeast to mammals. Additionally, mammals have gained an oligosaccharide structure-dependent folding cycle for glycoproteins. The molecular mechanisms of ERAD are also well conserved from yeast to mammals, but redundant expression of yeast orthologues in mammals has been encountered, particularly for components involved in recognition and processing of glycoproteins and components of the ER membrane complex involved in retrotranslocation and simultaneous ubiquitination of glycoproteins and non-glycoproteins. This may reflect an evolutionary consequence of increasing quantity or quality needs toward mammals. GENERAL SIGNIFICANCE The introduction of innovative genome editing technology into analysis of the mechanisms of mammalian ERAD, as exemplified here, will provide new insights into the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Satoshi Ninagawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan.
| | - Ginto George
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
13
|
Derlin-3 Is Required for Changes in ERAD Complex Formation under ER Stress. Int J Mol Sci 2020; 21:ijms21176146. [PMID: 32858914 PMCID: PMC7504720 DOI: 10.3390/ijms21176146] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/18/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
Endoplasmic reticulum (ER)-associated protein degradation (ERAD) is a quality control system that induces the degradation of ER terminally misfolded proteins. The ERAD system consists of complexes of multiple ER membrane-associated and luminal proteins that function cooperatively. We aimed to reveal the role of Derlin-3 in the ERAD system using the liver, pancreas, and kidney obtained from different mouse genotypes. We performed coimmunoprecipitation and sucrose density gradient centrifugation to unravel the dynamic nature of ERAD complexes. We observed that Derlin-3 is exclusively expressed in the pancreas, and its deficiency leads to the destabilization of Herp and accumulation of ERAD substrates. Under normal conditions, Complex-1a predominantly contains Herp, Derlin-2, HRD1, and SEL1L, and under ER stress, Complex-1b contains Herp, Derlin-3 (instead of Derlin-2), HRD1, and SEL1L. Complex-2 is upregulated under ER stress and contains Derlin-1, Derlin-2, p97, and VIMP. Derlin-3 deficiency suppresses the transition of Derlin-2 from Complex-1a to Complex-2 under ER stress. In the pancreas, Derlin-3 deficiency blocks Derlin-2 transition. In conclusion, the composition of ERAD complexes is tissue-specific and changes in response to ER stress in a Derlin-3-dependent manner. Derlin-3 may play a key role in changing ERAD complex compositions to overcome ER stress.
Collapse
|
14
|
Legros V, Jeannin P, Burlaud-Gaillard J, Chaze T, Gianetto QG, Butler-Browne G, Mouly V, Zoladek J, Afonso PV, Gonzàlez MN, Matondo M, Riederer I, Roingeard P, Gessain A, Choumet V, Ceccaldi PE. Differentiation-dependent susceptibility of human muscle cells to Zika virus infection. PLoS Negl Trop Dis 2020; 14:e0008282. [PMID: 32817655 PMCID: PMC7508361 DOI: 10.1371/journal.pntd.0008282] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/22/2020] [Accepted: 04/09/2020] [Indexed: 11/27/2022] Open
Abstract
Muscle cells are potential targets of many arboviruses, such as Ross River, Dengue, Sindbis, and chikungunya viruses, that may be involved in the physiopathological course of the infection. During the recent outbreak of Zika virus (ZIKV), myalgia was one of the most frequently reported symptoms. We investigated the susceptibility of human muscle cells to ZIKV infection. Using an in vitro model of human primary myoblasts that can be differentiated into myotubes, we found that myoblasts can be productively infected by ZIKV. In contrast, myotubes were shown to be resistant to ZIKV infection, suggesting a differentiation-dependent susceptibility. Infection was accompanied by a caspase-independent cytopathic effect, associated with paraptosis-like cytoplasmic vacuolization. Proteomic profiling was performed 24h and 48h post-infection in cells infected with two different isolates. Proteome changes indicate that ZIKV infection induces an upregulation of proteins involved in the activation of the Interferon type I pathway, and a downregulation of protein synthesis. This work constitutes the first observation of primary human muscle cells susceptibility to ZIKV infection, and differentiation-dependent restriction of infection from myoblasts to myotubes. Since myoblasts constitute the reservoir of stem cells involved in reparation/regeneration in muscle tissue, the infection of muscle cells and the viral-induced alterations observed here could have consequences in ZIKV infection pathogenesis. Muscle cells are potential targets of many arboviruses, such as Ross River, Dengue, Sindbis, and chikungunya viruses, and may be involved in the disease manifestation. During the recent outbreak of Zika virus (ZIKV), myalgia was one of the most frequently reported symptoms. We investigated the susceptibility of human muscle cells to ZIKV infection. Using an in vitro model of human muscle stem cells (myoblasts) that can be differentiated into differentiated muscle cells (myotubes), we found that myoblasts can be infected by ZIKV. In contrast, myotubes were shown to be resistant to ZIKV infection. Infection induced the death of infected cells. Protein levels 24h and 48h post-infection indicate that ZIKV infection induces an upregulation of proteins involved in the activation of the Interferon type I pathway, and a downregulation of protein synthesis. This work constitutes the first observation of primary human muscle cells susceptibility to ZIKV infection, muscle stem cells being susceptible while differentiated muscle cells are resistant. Since myoblasts constitute the reservoir of stem cells involved in reparation/regeneration in muscle tissue, the infection of muscle cells and the viral-induced alterations observed here could have consequences during ZIKV infection.
Collapse
Affiliation(s)
- Vincent Legros
- Unité Epidémiologie et Physiopathologie des Virus Oncogènes, Département de virologie, Institut Pasteur, Paris, France
- Université de Paris, Paris, France
- UMR CNRS 3569, Paris, France
| | - Patricia Jeannin
- Unité Epidémiologie et Physiopathologie des Virus Oncogènes, Département de virologie, Institut Pasteur, Paris, France
- Université de Paris, Paris, France
- UMR CNRS 3569, Paris, France
| | - Julien Burlaud-Gaillard
- INSERM U1259 & Plate Forme IBiSA de Microscopie Electronique, Université François Rabelais and CHRU, Tours, France
| | - Thibault Chaze
- Proteomics Platform, Mass Spectrometry for Biology Unit, USR 2000 IP CNRS, Institut Pasteur, Paris, France
| | - Quentin Giai Gianetto
- Proteomics Platform, Mass Spectrometry for Biology Unit, USR 2000 IP CNRS, Institut Pasteur, Paris, France
- Bioinformatics and Biostatistics Hub, C3BI, USR 3756 IP CNRS, Institut Pasteur, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Jim Zoladek
- Unité Epidémiologie et Physiopathologie des Virus Oncogènes, Département de virologie, Institut Pasteur, Paris, France
- Université de Paris, Paris, France
- UMR CNRS 3569, Paris, France
| | - Philippe V. Afonso
- Unité Epidémiologie et Physiopathologie des Virus Oncogènes, Département de virologie, Institut Pasteur, Paris, France
- Université de Paris, Paris, France
- UMR CNRS 3569, Paris, France
| | - Mariela-Natacha Gonzàlez
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, USR 2000 IP CNRS, Institut Pasteur, Paris, France
| | - Ingo Riederer
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | - Philippe Roingeard
- INSERM U1259 & Plate Forme IBiSA de Microscopie Electronique, Université François Rabelais and CHRU, Tours, France
| | - Antoine Gessain
- Unité Epidémiologie et Physiopathologie des Virus Oncogènes, Département de virologie, Institut Pasteur, Paris, France
- Université de Paris, Paris, France
- UMR CNRS 3569, Paris, France
| | - Valérie Choumet
- Unité Environnement et Risques Infectieux, Département de santé globale, Institut Pasteur, Paris, France
- * E-mail: (VC); (PEC)
| | - Pierre-Emmanuel Ceccaldi
- Unité Epidémiologie et Physiopathologie des Virus Oncogènes, Département de virologie, Institut Pasteur, Paris, France
- Université de Paris, Paris, France
- UMR CNRS 3569, Paris, France
- * E-mail: (VC); (PEC)
| |
Collapse
|
15
|
Fenech EJ, Lari F, Charles PD, Fischer R, Laétitia-Thézénas M, Bagola K, Paton AW, Paton JC, Gyrd-Hansen M, Kessler BM, Christianson JC. Interaction mapping of endoplasmic reticulum ubiquitin ligases identifies modulators of innate immune signalling. eLife 2020; 9:e57306. [PMID: 32614325 PMCID: PMC7332293 DOI: 10.7554/elife.57306] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/11/2020] [Indexed: 12/25/2022] Open
Abstract
Ubiquitin ligases (E3s) embedded in the endoplasmic reticulum (ER) membrane regulate essential cellular activities including protein quality control, calcium flux, and sterol homeostasis. At least 25 different, transmembrane domain (TMD)-containing E3s are predicted to be ER-localised, but for most their organisation and cellular roles remain poorly defined. Using a comparative proteomic workflow, we mapped over 450 protein-protein interactions for 21 stably expressed, full-length E3s. Bioinformatic analysis linked ER-E3s and their interactors to multiple homeostatic, regulatory, and metabolic pathways. Among these were four membrane-embedded interactors of RNF26, a polytopic E3 whose abundance is auto-regulated by ubiquitin-proteasome dependent degradation. RNF26 co-assembles with TMEM43, ENDOD1, TMEM33 and TMED1 to form a complex capable of modulating innate immune signalling through the cGAS-STING pathway. This RNF26 complex represents a new modulatory axis of STING and innate immune signalling at the ER membrane. Collectively, these data reveal the broad scope of regulation and differential functionalities mediated by ER-E3s for both membrane-tethered and cytoplasmic processes.
Collapse
Affiliation(s)
- Emma J Fenech
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Federica Lari
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Philip D Charles
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
| | - Roman Fischer
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
| | - Marie Laétitia-Thézénas
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
| | - Katrin Bagola
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of AdelaideAdelaideAustralia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of AdelaideAdelaideAustralia
| | - Mads Gyrd-Hansen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Benedikt M Kessler
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of OxfordOxfordUnited Kingdom
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - John C Christianson
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Botnar Research CentreOxfordUnited Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Botnar Research CentreOxfordUnited Kingdom
| |
Collapse
|
16
|
Shenkman M, Lederkremer GZ. Compartmentalization and Selective Tagging for Disposal of Misfolded Glycoproteins. Trends Biochem Sci 2019; 44:827-836. [PMID: 31133362 DOI: 10.1016/j.tibs.2019.04.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 01/08/2023]
Abstract
The ability of mammalian cells to correctly identify and degrade misfolded secretory proteins, most of them bearing N-glycans, is crucial for their correct function and survival. An inefficient disposal mechanism results in the accumulation of misfolded proteins and consequent endoplasmic reticulum (ER) stress. N-glycan processing creates a code that reveals the folding status of each molecule, enabling continued folding attempts or targeting of the doomed glycoprotein for disposal. We review here the main steps involved in the accurate processing of unfolded glycoproteins. We highlight recent data suggesting that the processing is not stochastic, but that there is selective accelerated glycan trimming on misfolded glycoprotein molecules.
Collapse
Affiliation(s)
- Marina Shenkman
- School of Molecular Cell Biology and Biotechnology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerardo Z Lederkremer
- School of Molecular Cell Biology and Biotechnology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
17
|
Chen F, Wen X, Lin P, Chen H, Wang A, Jin Y. HERP depletion inhibits zearalenone-induced apoptosis through autophagy activation in mouse ovarian granulosa cells. Toxicol Lett 2018; 301:1-10. [PMID: 30394307 DOI: 10.1016/j.toxlet.2018.10.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/25/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
HERP is an endoplasmic reticulum (ER) membrane protein and is strongly induced by stress conditions. A recent study has indicated that HERP cooperates in apoptosis during zearalenone (ZEA) treatment. However, regulatory mechanisms and the role of HERP in ZEA-induced apoptosis remain elusive in ovarian granulosa cells. In this study, MTT and flow cytometry assays demonstrated that ZEA gradually decreased cell viability and increased apoptosis in granulosa cells in a dose-dependent manner. Western blot analysis showed that ZEA significantly activated autophagy by upregulating LC3-II. Chloroquine (CQ) significantly increased LC3-II and induced granulosa cell apoptosis. Moreover, Western blot analysis showed that ZEA inhibited the mTOR and ERK1/2 signaling pathways. Furthermore, we found that ZEA activated ER stress by upregulating the ER stress-related proteins GRP78, HERP and CHOP. 4-PBA significantly decreased GRP78, HERP, CHOP and LC3-II. In addition, knockdown of HERP (shHERP) significantly protected ovarian granulosa cells from apoptosis induced by ZEA. We found that HERP depletion activated autophagy and ERK1/2 signaling pathways, while it inhibited the mTOR and caspase-dependent mitochondrial signaling pathways. In summary, autophagy and ER stress cooperated in apoptosis induced by ZEA; HERP depletion inhibits ZEA-induced apoptosis of ovarian granulosa cells through autophagy activation and apoptotic pathway inhibition.
Collapse
Affiliation(s)
- Fenglei Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China; College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Xin Wen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Huatao Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
18
|
Maeda T, Fujita Y, Tanabe-Fujimura C, Zou K, Liu J, Liu S, Kikuchi K, Shen X, Nakajima T, Komano H. An E3 Ubiquitin Ligase, Synoviolin, Is Involved in the Degradation of Homocysteine-Inducible Endoplasmic Reticulum Protein. Biol Pharm Bull 2018; 41:915-919. [PMID: 29863080 DOI: 10.1248/bpb.b18-00015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Homocysteine-inducible endoplasmic reticulum (ER) protein (Herp) is an ER stress-inducible membrane protein involved in ER-associated degradation. Herp expression is maintained at low levels through a strict regulatory mechanism, but the details of this mechanism and the reasons why Herp expression is restricted in this manner remain unclear. Here, we show that Herp degradation involves synoviolin, an ER-resident E3 ubiquitin ligase. Herp protein levels were found to be markedly elevated in synoviolin-null cells, and Herp expression decreased when synoviolin was overexpressed. However, the lysine residues of Herp, which are ubiquitinated by E3 ubiquitin ligase, were not sufficient for regulation of Herp degradation. These results suggest that Herp degradation is mediated via synoviolin and that Herp ubiquitination involves amino acids other than lysine.
Collapse
Affiliation(s)
- Tomoji Maeda
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | - Yu Fujita
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | | | - Kun Zou
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | - Junjun Liu
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | - Shuyu Liu
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | - Kota Kikuchi
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | - Xuefeng Shen
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| | | | - Hiroto Komano
- Department of Neuroscience, School of Pharmacy, Iwate Medical University
| |
Collapse
|
19
|
Barrera MJ, Aguilera S, Castro I, González S, Carvajal P, Molina C, Hermoso MA, González MJ. Endoplasmic reticulum stress in autoimmune diseases: Can altered protein quality control and/or unfolded protein response contribute to autoimmunity? A critical review on Sjögren's syndrome. Autoimmun Rev 2018; 17:796-808. [PMID: 29890347 DOI: 10.1016/j.autrev.2018.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022]
Abstract
For many years, researchers in the field of autoimmunity have focused on the role of the immune components in the etiopathogenesis of autoimmune diseases. However, some studies have demonstrated the importance of target tissues in their pathogenesis and the breach of immune tolerance. The immune system as well as target tissue cells (plasmatic, β-pancreatic, fibroblast-like synoviocytes, thyroid follicular and epithelial cells of the lachrymal glands, salivary glands, intestine, bronchioles and renal tubules) share the characteristic of secretory cells with an extended endoplasmic reticulum (ER). The function of these cells depends considerably on a normal ER function and calcium homeostasis, so they can produce and secrete their main components, which include glycoproteins involved in antigenic presentation such as major histocompatibility complex (MHC) class I and II. All these proteins are synthesized and modified in the ER, and for this reason disturbances in the normal functions of this organelle such as protein folding, protein quality control, calcium homeostasis and redox balance, promote accumulation of unfolded or misfolded proteins, a condition known as ER stress. Autoimmune diseases are characterized by inflammation, which has been associated with an ER stress condition. Interestingly, patients with these diseases contain circulating auto-antibodies against chaperone proteins (such as Calnexin and GRP94), thus affecting the folding and assembly of MHC class I and II glycoproteins and their loading with peptide. The main purpose of this article is to review the involvement of the protein quality control and unfolded protein response (UPR) in the ER protein homeostasis (proteostasis) and their alterations in autoimmune diseases. In addition, we describe the interaction between ER stress and inflammation and evidences are shown of how autoimmune diseases are associated with an ER stress condition, with a special emphasis on the second most prevalent autoimmune rheumatic disease, Sjögren's syndrome.
Collapse
Affiliation(s)
- María-José Barrera
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Aguilera
- Departamento de Reumatología, Clínica INDISA, Santiago, Chile
| | - Isabel Castro
- Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio González
- Escuela de Odontología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Patricia Carvajal
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Claudio Molina
- Escuela de Postgrado, Facultad de Odontología, Universidad San Sebastián, Santiago, Chile
| | - Marcela A Hermoso
- Programa de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María-Julieta González
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
20
|
Norisada J, Fujimura K, Amaya F, Kohno H, Hirata Y, Oh-hashi K. Application of NanoBiT for Monitoring Dimerization of the Null Hong Kong Variant of α-1-Antitrypsin, NHK, in Living Cells. Mol Biotechnol 2018; 60:539-549. [DOI: 10.1007/s12033-018-0092-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
21
|
Américo-Da-Silva L, Diaz J, Bustamante M, Mancilla G, Oyarzún I, Verdejo HE, Quiroga C. A new role for HERPUD1 and ERAD activation in osteoblast differentiation and mineralization. FASEB J 2018; 32:4681-4695. [PMID: 29570393 DOI: 10.1096/fj.201701229rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bone integrity depends on a finely tuned balance between bone synthesis by osteoblasts and resorption by osteoclasts. The secretion capacity of mature osteoblasts requires strict control of proteostasis. Endoplasmic reticulum-associated degradation (ERAD) prevents the accumulation of unfolded ER proteins via dislocation to the cytosol and degradation by the proteasome. The ER membrane protein, homocysteine-inducible endoplasmic reticulum protein with ubiquitin-like domain 1 (HERPUD1), is a key component of the ERAD multiprotein complex which helps to stabilize the complex and facilitate the efficient degradation of unfolded proteins. HERPUD1 expression is strongly up-regulated by the unfolded protein response and cellular stress. The aim of the current study was to establish whether HERPUD1 and ERAD play roles in osteoblast differentiation and maturation. We evaluated preosteoblastic MC3T3-E1 cell and primary rat osteoblast differentiation by measuring calcium deposit levels, alkaline phosphatase activity, and runt-related transcription factor 2 and osterix expression. We found that ERAD and proteasomal degradation were activated and that HERPUD1 expression was increased as osteoblast differentiation progressed. The absence of HERPUD1 blocked osteoblast mineralization in vitro and significantly reduced alkaline phosphatase activity. In contrast, HERPUD1 overexpression activated the osteoblast differentiation program. Our results demonstrate that HERPUD1 and ERAD are important for the activation of the osteoblast maturation program and may be useful new targets for elucidating bone physiology.-Américo-Da-Silva, L., Diaz, J., Bustamante, M., Mancilla, G., Oyarzún, I., Verdejo, H. E., Quiroga, C. A new role for HERPUD1 and ERAD activation in osteoblast differentiation and mineralization.
Collapse
Affiliation(s)
- Luan Américo-Da-Silva
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jheimmy Diaz
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mario Bustamante
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Georthan Mancilla
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ingrid Oyarzún
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo E Verdejo
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Clara Quiroga
- División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Universidad de Chile and Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
22
|
Luo H, Cao L, Liang X, Du A, Peng T, Li H. Herp Promotes Degradation of Mutant Huntingtin: Involvement of the Proteasome and Molecular Chaperones. Mol Neurobiol 2018; 55:7652-7668. [PMID: 29430620 DOI: 10.1007/s12035-018-0900-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/09/2018] [Indexed: 01/18/2023]
Abstract
In neurodegenerative diseases, pathogenic proteins tend to misfold and form aggregates that are difficult to remove and able to induce excessive endoplasmic reticulum (ER) stress, leading to neuronal injury and apoptosis. Homocysteine-induced endoplasmic reticulum protein (Herp), an E3 ubiquitin ligase, is an important early marker of ER stress and is involved in the ubiquitination and degradation of many neurodegenerative proteins. However, in Huntington's disease (HD), a typical polyglutamine disease, whether Herp is also involved in the metabolism and degradation of the pathogenic protein, mutant huntingtin, has not been reported. Therefore, we studied the relationship between Herp and N-terminal fragments of huntingtin (HttN-20Q and HttN-160Q). We found that Herp was able to bind to the overexpressed Htt N-terminal, and this interaction was enhanced by expansion of the polyQ fragment. Confocal microscopy demonstrated that Herp was co-localized with the HttN-160Q aggregates in the cytoplasm and tightly surrounded the aggregates. Overexpression of Herp significantly decreased the amount of soluble and insoluble HttN-160Q, promoted its ubiquitination, and inhibited its cytotoxicity. In contrast, knockdown of Herp resulted in more HttN-160Q protein, less ubiquitination, and stronger cytotoxicity. Inhibition of the autophagy-lysosomal pathway (ALP) had no effect on the function of Herp. However, blocking the ubiquitin-proteasome pathway (UPP) inhibited the reduction in soluble HttN-160Q caused by Herp. Interestingly, blocking the UPP did not weaken the ability of Herp to reduce HttN-160Q aggregates. Deletions of the N-terminal of Herp weakened its ability to inhibit HttN-160Q aggregation but did not result in a significant increase in its soluble form. However, loss of the C-terminal led to a significant increase in soluble HttN-160Q, but Herp still maintained the ability to inhibit aggregate formation. We further found that the expression level of Herp was significantly increased in HD animal and cell models. Our findings suggest that Herp is a newly identified huntingtin-interacting protein that is able to reduce the cytotoxicity of mutant huntingtin by inhibiting its aggregation and promoting its degradation. The N-terminal of Herp serves as the molecular chaperone to inhibit protein aggregation, while its C-terminal functions as an E3 ubiquitin ligase to promote the degradation of misfolded proteins through the UPP. Increased expression of Herp in HD models may be a pro-survival mechanism under stress.
Collapse
Affiliation(s)
- Huanhuan Luo
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.,Department of Histology and Embryology, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Liying Cao
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xuan Liang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ana Du
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ting Peng
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - He Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China. .,Department of Histology and Embryology, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
| |
Collapse
|
23
|
Wang T, Wang B, Huang H, Zhang C, Zhu Y, Pei B, Cheng C, Sun L, Wang J, Jin Q, Zhao Z. Enterovirus 71 protease 2Apro and 3Cpro differentially inhibit the cellular endoplasmic reticulum-associated degradation (ERAD) pathway via distinct mechanisms, and enterovirus 71 hijacks ERAD component p97 to promote its replication. PLoS Pathog 2017; 13:e1006674. [PMID: 28985237 PMCID: PMC5650186 DOI: 10.1371/journal.ppat.1006674] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/20/2017] [Accepted: 09/28/2017] [Indexed: 11/19/2022] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) is an important function for cellular homeostasis. The mechanism of how picornavirus infection interferes with ERAD remains unclear. In this study, we demonstrated that enterovirus 71 (EV71) infection significantly inhibits cellular ERAD by targeting multiple key ERAD molecules with its proteases 2Apro and 3Cpro using different mechanisms. Ubc6e was identified as the key E2 ubiquitin-conjugating enzyme in EV71 disturbed ERAD. EV71 3Cpro cleaves Ubc6e at Q219G, Q260S, and Q273G. EV71 2Apro mainly inhibits the de novo synthesis of key ERAD molecules Herp and VIMP at the protein translational level. Herp differentially participates in the degradation of different glycosylated ERAD substrates α-1 antitrypsin Null Hong Kong (NHK) and the C-terminus of sonic hedgehog (SHH-C) via unknown mechanisms. p97 was identified as a host factor in EV71 replication; it redistributed and co-exists with the viral protein and other known replication-related molecules in EV71-induced replication organelles. Electron microscopy and multiple-color confocal assays also showed that EV71-induced membranous vesicles were closely associated with the endoplasmic reticulum (ER), and the ER membrane molecule RTN3 was redistributed to the viral replication complex during EV71 infection. Therefore, we propose that EV71 rearranges ER membranes and hijacks p97 from cellular ERAD to benefit its replication. These findings add to our understanding of how viruses disturb ERAD and provide potential anti-viral targets for EV71 infection.
Collapse
Affiliation(s)
- Tao Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Bei Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - He Huang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Chongyang Zhang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Yuanmei Zhu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Bin Pei
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Chaofei Cheng
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Lei Sun
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, PR China
| | - Jianwei Wang
- MOH Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, IPB, CAMS-Fondation Mérieux, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- * E-mail: (JWW); (QJ); (ZDZ)
| | - Qi Jin
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- * E-mail: (JWW); (QJ); (ZDZ)
| | - Zhendong Zhao
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- Center of Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- CAMS-Oxford University International Center for Translational Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- * E-mail: (JWW); (QJ); (ZDZ)
| |
Collapse
|
24
|
Ge M, Luo Z, Qiao Z, Zhou Y, Cheng X, Geng Q, Cai Y, Wan P, Xiong Y, Liu F, Wu K, Liu Y, Wu J. HERP Binds TBK1 To Activate Innate Immunity and Repress Virus Replication in Response to Endoplasmic Reticulum Stress. THE JOURNAL OF IMMUNOLOGY 2017; 199:3280-3292. [DOI: 10.4049/jimmunol.1700376] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022]
|
25
|
Schulz J, Avci D, Queisser MA, Gutschmidt A, Dreher LS, Fenech EJ, Volkmar N, Hayashi Y, Hoppe T, Christianson JC. Conserved cytoplasmic domains promote Hrd1 ubiquitin ligase complex formation for ER-associated degradation (ERAD). J Cell Sci 2017; 130:3322-3335. [PMID: 28827405 DOI: 10.1242/jcs.206847] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022] Open
Abstract
The mammalian ubiquitin ligase Hrd1 is the central component of a complex facilitating degradation of misfolded proteins during the ubiquitin-proteasome-dependent process of ER-associated degradation (ERAD). Hrd1 associates with cofactors to execute ERAD, but their roles and how they assemble with Hrd1 are not well understood. Here, we identify crucial cofactor interaction domains within Hrd1 and report a previously unrecognised evolutionarily conserved segment within the intrinsically disordered cytoplasmic domain of Hrd1 (termed the HAF-H domain), which engages complementary segments in the cofactors FAM8A1 and Herp (also known as HERPUD1). This domain is required by Hrd1 to interact with both FAM8A1 and Herp, as well as to assemble higher-order Hrd1 complexes. FAM8A1 enhances binding of Herp to Hrd1, an interaction that is required for ERAD. Our findings support a model of Hrd1 complex formation, where the Hrd1 cytoplasmic domain and FAM8A1 have a central role in the assembly and activity of this ERAD machinery.
Collapse
Affiliation(s)
- Jasmin Schulz
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Dönem Avci
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Markus A Queisser
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Aljona Gutschmidt
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Lena-Sophie Dreher
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Emma J Fenech
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Norbert Volkmar
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Yuki Hayashi
- Department of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Thorsten Hoppe
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - John C Christianson
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
26
|
Joly P, Vignaud H, Di Martino J, Ruiz M, Garin R, Restier L, Belmalih A, Marchal C, Cullin C, Arveiler B, Fergelot P, Gitler AD, Lachaux A, Couthouis J, Bouchecareilh M. ERAD defects and the HFE-H63D variant are associated with increased risk of liver damages in Alpha 1-Antitrypsin Deficiency. PLoS One 2017; 12:e0179369. [PMID: 28617828 PMCID: PMC5472284 DOI: 10.1371/journal.pone.0179369] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/30/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The most common and severe disease causing allele of Alpha 1-Antitrypsin Deficiency (1ATD) is Z-1AT. This protein aggregates in the endoplasmic reticulum, which is the main cause of liver disease in childhood. Based on recent evidences and on the frequency of liver disease occurrence in Z-1AT patients, it seems that liver disease progression is linked to still unknown genetic factors. METHODS We used an innovative approach combining yeast genetic screens with next generation exome sequencing to identify and functionally characterize the genes involved in 1ATD associated liver disease. RESULTS Using yeast genetic screens, we identified HRD1, an Endoplasmic Reticulum Associated Degradation (ERAD) associated protein, as an inducer of Z-mediated toxicity. Whole exome sequencing of 1ATD patients resulted in the identification of two variants associated with liver damages in Z-1AT homozygous cases: HFE H63D and HERPUD1 R50H. Functional characterization in Z-1AT model cell lines demonstrated that impairment of the ERAD machinery combined with the HFE H63D variant expression decreased both cell proliferation and cell viability, while Unfolded Protein Response (UPR)-mediated cell death was hyperstimulated. CONCLUSION This powerful experimental pipeline allowed us to identify and functionally validate two genes involved in Z-1AT-mediated severe liver toxicity. This pilot study moves forward our understanding on genetic modifiers involved in 1ATD and highlights the UPR pathway as a target for the treatment of liver diseases associated with 1ATD. Finally, these findings support a larger scale screening for HERPUD1 R50H and HFE H63D variants in the sub-group of 1ATD patients developing significant chronic hepatic injuries (hepatomegaly, chronic cholestasis, elevated liver enzymes) and at risk developing liver cirrhosis.
Collapse
Affiliation(s)
- Philippe Joly
- University Lyon - University Claude Bernard Lyon 1 - EA 7424 – Inter-university Laboratory of Human Movement Science, Villeurbanne, France
- Laboratoire de Biochimie et biologie moléculaire Grand-Est, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Hélène Vignaud
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
| | - Julie Di Martino
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
- INSERM, University Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux, France
| | - Mathias Ruiz
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Roman Garin
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Lioara Restier
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Abdelouahed Belmalih
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Christelle Marchal
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
| | - Christophe Cullin
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
| | - Benoit Arveiler
- University Bordeaux, INSERM U1211, Laboratoire Maladies Rares, Génétique et Métabolisme (MRGM), Bordeaux, France
| | - Patricia Fergelot
- University Bordeaux, INSERM U1211, Laboratoire Maladies Rares, Génétique et Métabolisme (MRGM), Bordeaux, France
| | - Aaron D. Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Alain Lachaux
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Lyon, Lyon, France
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Marion Bouchecareilh
- CNRS, University Bordeaux, UMR5095 Institut de Biochimie et Génétique Cellulaires, Bordeaux, France
- INSERM, University Bordeaux, UMR1053 Bordeaux Research In Translational Oncology, BaRITOn, Bordeaux, France
- * E-mail:
| |
Collapse
|
27
|
Hwang J, Walczak CP, Shaler TA, Olzmann JA, Zhang L, Elias JE, Kopito RR. Characterization of protein complexes of the endoplasmic reticulum-associated degradation E3 ubiquitin ligase Hrd1. J Biol Chem 2017; 292:9104-9116. [PMID: 28411238 PMCID: PMC5454095 DOI: 10.1074/jbc.m117.785055] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
Hrd1 is the core structural component of a large endoplasmic reticulum membrane-embedded protein complex that coordinates the destruction of folding-defective proteins in the early secretory pathway. Defining the composition, dynamics, and ultimately, the structure of the Hrd1 complex is a crucial step in understanding the molecular basis of glycoprotein quality control but has been hampered by the lack of suitable techniques to interrogate this complex under native conditions. In this study we used genome editing to generate clonal HEK293 (Hrd1.KI) cells harboring a homozygous insertion of a small tandem affinity tag knocked into the endogenous Hrd1 locus. We found that steady-state levels of tagged Hrd1 in these cells are indistinguishable from those of Hrd1 in unmodified cells and that the tagged variant is functional in supporting the degradation of well characterized luminal and membrane substrates. Analysis of detergent-solubilized Hrd1.KI cells indicates that the composition and stoichiometry of Hrd1 complexes are strongly influenced by Hrd1 expression levels. Analysis of affinity-captured Hrd1 complexes from these cells by size-exclusion chromatography, immunodepletion, and absolute quantification mass spectrometry identified two major high-molecular-mass complexes with distinct sets of interacting proteins and variable stoichiometries, suggesting a hitherto unrecognized heterogeneity in the functional units of Hrd1-mediated protein degradation.
Collapse
Affiliation(s)
| | | | | | | | - Lichao Zhang
- Chemical and Systems Biology, Stanford University, Stanford, California 94305 and
| | - Joshua E Elias
- Chemical and Systems Biology, Stanford University, Stanford, California 94305 and
| | | |
Collapse
|
28
|
The NF-κB-dependent and -independent transcriptome and chromatin landscapes of human coronavirus 229E-infected cells. PLoS Pathog 2017; 13:e1006286. [PMID: 28355270 PMCID: PMC5386326 DOI: 10.1371/journal.ppat.1006286] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/10/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Coronavirus replication takes place in the host cell cytoplasm and triggers inflammatory gene expression by poorly characterized mechanisms. To obtain more insight into the signals and molecular events that coordinate global host responses in the nucleus of coronavirus-infected cells, first, transcriptome dynamics was studied in human coronavirus 229E (HCoV-229E)-infected A549 and HuH7 cells, respectively, revealing a core signature of upregulated genes in these cells. Compared to treatment with the prototypical inflammatory cytokine interleukin(IL)-1, HCoV-229E replication was found to attenuate the inducible activity of the transcription factor (TF) NF-κB and to restrict the nuclear concentration of NF-κB subunits by (i) an unusual mechanism involving partial degradation of IKKβ, NEMO and IκBα and (ii) upregulation of TNFAIP3 (A20), although constitutive IKK activity and basal TNFAIP3 expression levels were shown to be required for efficient virus replication. Second, we characterized actively transcribed genomic regions and enhancers in HCoV-229E-infected cells and systematically correlated the genome-wide gene expression changes with the recruitment of Ser5-phosphorylated RNA polymerase II and prototypical histone modifications (H3K9ac, H3K36ac, H4K5ac, H3K27ac, H3K4me1). The data revealed that, in HCoV-infected (but not IL-1-treated) cells, an extensive set of genes was activated without inducible p65 NF-κB being recruited. Furthermore, both HCoV-229E replication and IL-1 were shown to upregulate a small set of genes encoding immunomodulatory factors that bind p65 at promoters and require IKKβ activity and p65 for expression. Also, HCoV-229E and IL-1 activated a common set of 440 p65-bound enhancers that differed from another 992 HCoV-229E-specific enhancer regions by distinct TF-binding motif combinations. Taken together, the study shows that cytoplasmic RNA viruses fine-tune NF-κB signaling at multiple levels and profoundly reprogram the host cellular chromatin landscape, thereby orchestrating the timely coordinated expression of genes involved in multiple signaling, immunoregulatory and metabolic processes. Coronaviruses are major human and animal pathogens. They belong to a family of plus-strand RNA viruses that have extremely large genomes and encode a variety of proteins involved in virus-host interactions. The four common coronaviruses (HCoV-229E, NL63, OC43, HKU1) cause mainly upper respiratory tract infections, while zoonotic coronaviruses (SARS-CoV and MERS-CoV) cause severe lung disease, including acute respiratory distress syndrome (ARDS). The molecular basis for this fundamentally different pathology is incompletely understood. Our study provides a genome-wide investigation of epigenetic changes occurring in response to HCoV-229E. We identify at high resolution a large number of regulatory regions in the genome of infected cells that coordinate de novo gene transcription. Many of these genes have immunomodulatory functions and, most likely, contribute to limiting viral replication, while other factors may promote viral replication. The study provides an intriguing example of a virus that completes its entire life cycle in the cytoplasm while sending multiple signals to the nuclear chromatin compartment to adjust the host cell repertoire of transcribed genes. The approach taken in this study is expected to provide a suitable framework for future studies aimed at dissecting and comparing host responses to representative coronaviruses with different pathogenic potential in humans.
Collapse
|
29
|
Ding W, Chen R, Wu C, Chen W, Zhang H, Fan X, Wang H, Ji Y, Xie L, Ning X, Shen L. Increased expression of HERPUD1 involves in neuronal apoptosis after intracerebral hemorrhage. Brain Res Bull 2016; 128:40-47. [PMID: 27871950 DOI: 10.1016/j.brainresbull.2016.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/13/2016] [Accepted: 11/16/2016] [Indexed: 10/20/2022]
Abstract
Homocysteine-inducible endoplasmic reticulum stress-inducible ubiquitin-like domain member 1 protein (HERPUD1) is involved in endoplasmic reticulum stress response. Immense amounts of research showed HERPUD1 plays multiple roles in various models. In this work, we explored the role of HERPUD1 during the pathophysiological processes of intracerebral hemorrhage (ICH). Rat ICH model was established and verified by behavioral test. Western blot and immunohistochemistry revealed a significant up-regulation of HERPUD1 expression around the hematoma after ICH. Besides, the expression of cytochrome c (cyt c) and active caspase-3 increased accompanied to HERPUD1 expression. Double-labeled immunofluorescence indicated HERPUD1 mainly colocalized with neurons. Further study showed HERPUD1 silence brought about up-regulation of apoptosis markers including cyt c and active caspase-3 coupled with increased cell apoptosis in vitro model. All these findings suggested that HERPUD1 might play a protective role in ICH-induced neuronal apoptosis in rat models.
Collapse
Affiliation(s)
- Wensen Ding
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Rongrong Chen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Chunshuai Wu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Weihai Chen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Hongtian Zhang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Xingjuan Fan
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Hongmei Wang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Yuteng Ji
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Lili Xie
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Xiaojin Ning
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Lihua Shen
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
30
|
Jeong H, Sim HJ, Song EK, Lee H, Ha SC, Jun Y, Park TJ, Lee C. Crystal structure of SEL1L: Insight into the roles of SLR motifs in ERAD pathway. Sci Rep 2016; 6:20261. [PMID: 27064360 PMCID: PMC4746701 DOI: 10.1038/srep20261] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/31/2015] [Indexed: 11/09/2022] Open
Abstract
Terminally misfolded proteins are selectively recognized and cleared by the endoplasmic reticulum-associated degradation (ERAD) pathway. SEL1L, a component of the ERAD machinery, plays an important role in selecting and transporting ERAD substrates for degradation. We have determined the crystal structure of the mouse SEL1L central domain comprising five Sel1-Like Repeats (SLR motifs 5 to 9; hereafter called SEL1Lcent). Strikingly, SEL1Lcent forms a homodimer with two-fold symmetry in a head-to-tail manner. Particularly, the SLR motif 9 plays an important role in dimer formation by adopting a domain-swapped structure and providing an extensive dimeric interface. We identified that the full-length SEL1L forms a self-oligomer through the SEL1Lcent domain in mammalian cells. Furthermore, we discovered that the SLR-C, comprising SLR motifs 10 and 11, of SEL1L directly interacts with the N-terminus luminal loops of HRD1. Therefore, we propose that certain SLR motifs of SEL1L play a unique role in membrane bound ERAD machinery.
Collapse
Affiliation(s)
- Hanbin Jeong
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Hyo Jung Sim
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Eun Kyung Song
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Hakbong Lee
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Sung Chul Ha
- Pohang Accelerator Laboratory, Pohang University of Science and Technology, Pohang, Kyungbuk 37673, Korea
| | - Youngsoo Jun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea
| | - Tae Joo Park
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST-gil, Ulsan 44919, Republic of Korea
| | - Changwook Lee
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST-gil, Ulsan 44919, Republic of Korea
| |
Collapse
|
31
|
Chen F, Wang N, Yang D, Wen X, Mahmoud TN, Zhou D, Tang K, Lin P, Wang A, Jin Y. Herp depletion arrests the S phase of the cell cycle and increases estradiol synthesis in mouse granulosa cells. J Reprod Dev 2016; 62:159-66. [PMID: 26781490 PMCID: PMC4848573 DOI: 10.1262/jrd.2015-120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The endoplasmic reticulum (ER) stress response has been implicated in the development, atresia and luteinization of ovarian follicles. However, there have been few reports concerning the role of Herp, an ER stress-induced protein, in follicular development. The present study aims to detect the distribution and cyclic variations of Herp during the estrous cycle and to reveal the roles of Herp in regulating the cell cycle, apoptosis and steroid hormone biosynthesis in mouse granulosa cells. In this study, immunohistochemistry staining showed that Herp expression was primarily in the granulosa cells and oocytes. Furthermore, we constructed recombinant lentiviral vectors for Herp short hairpin interfering RNA (shRNA) expression; immunofluorescence staining, real-time quantitative PCR (RT-qPCR) and western blot analysis revealed that Herp was successfully knocked down. Flow cytometry showed that knockdown of Herp arrested granulosa cells at the S phase of the cell cycle. More importantly, ELISA analysis revealed that Herp knockdown significantly upregulated the concentration of estradiol (E2) in the culture supernatants. RT-qPCR was performed to determine the regulatory mechanism of Herp knockdown in the cell cycle, and in steroid synthesis, RT-qPCR analysis revealed that Herp knockdown upregulated the mRNA expression of steroidogenic enzymes (Cyp19a1) and downregulated metabolic enzymes (Cyp1b1) and cell cycle factors (cyclin A1, cyclin B1 and cyclin D2). These results suggest that Herp may regulate the cell cycle and hormone secretions in mouse granulosa cells. The present study helps to elucidate the physiological functions of Herp as they relate to reproduction.
Collapse
Affiliation(s)
- Fenglei Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Shaanxi 712100, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Paredes F, Parra V, Torrealba N, Navarro-Marquez M, Gatica D, Bravo-Sagua R, Troncoso R, Pennanen C, Quiroga C, Chiong M, Caesar C, Taylor WR, Molgó J, San Martin A, Jaimovich E, Lavandero S. HERPUD1 protects against oxidative stress-induced apoptosis through downregulation of the inositol 1,4,5-trisphosphate receptor. Free Radic Biol Med 2016; 90:206-18. [PMID: 26616647 PMCID: PMC4710961 DOI: 10.1016/j.freeradbiomed.2015.11.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 11/17/2015] [Accepted: 11/20/2015] [Indexed: 11/27/2022]
Abstract
Homocysteine-inducible, endoplasmic reticulum (ER) stress-inducible, ubiquitin-like domain member 1 (HERPUD1), an ER resident protein, is upregulated in response to ER stress and Ca(2+) homeostasis deregulation. HERPUD1 exerts cytoprotective effects in various models, but its role during oxidative insult remains unknown. The aim of this study was to investigate whether HERPUD1 contributes to cytoprotection in response to redox stress and participates in mediating stress-dependent signaling pathways. Our data showed that HERPUD1 protein levels increased in HeLa cells treated for 30 min with H2O2 or angiotensin II and in aortic tissue isolated from mice treated with angiotensin II for 3 weeks. Cell death was higher in HERPUD1 knockdown (sh-HERPUD1) HeLa cells treated with H2O2 in comparison with control (sh-Luc) HeLa cells. This effect was abolished by the intracellular Ca(2+) chelating agent BAPTA-AM or the inositol 1,4,5-trisphosphate receptor (ITPR) antagonist xestospongin B, suggesting that the response to H2O2 was dependent on intracellular Ca(2+) stores and the ITPR. Ca(2+) kinetics showed that sh-HERPUD1 HeLa cells exhibited greater and more sustained cytosolic and mitochondrial Ca(2+) increases than sh-Luc HeLa cells. This higher sensitivity of sh-HERPUD1 HeLa cells to H2O2 was prevented with the mitochondrial permeability transition pore inhibitor cyclosporine A. We concluded that the HERPUD1-mediated cytoprotective effect against oxidative stress depends on the ITPR and Ca(2+) transfer from the ER to mitochondria.
Collapse
Affiliation(s)
- Felipe Paredes
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Valentina Parra
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Natalia Torrealba
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Mario Navarro-Marquez
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Damian Gatica
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Rodrigo Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile; Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago, Chile
| | - Christian Pennanen
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Clara Quiroga
- ACCDiS, Cardiovascular Diseases Division, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile
| | - Christa Caesar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - W Robert Taylor
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, USA; Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jordi Molgó
- Institut des Neurosciences Paris-Saclay, UMR 9197, 91190 Gif sur Yvette, France
| | - Alejandra San Martin
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la Célula, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, 838049 Santiago, Chile; Centro de Estudios Moleculares de la Célula, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
33
|
Chen F, Lin P, Wang N, Yang D, Wen X, Zhou D, Wang A, Jin Y. Herp depletion inhibits zearalenone-induced cell death in RAW 264.7 macrophages. Toxicol In Vitro 2015; 32:115-22. [PMID: 26723276 DOI: 10.1016/j.tiv.2015.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 11/26/2015] [Accepted: 12/18/2015] [Indexed: 12/14/2022]
Abstract
Herp is an endoplasmic reticulum (ER) membrane protein and strongly induced by the ER stress that not only participates in the unfolded protein response (UPR) under the ER stress, but also in cell autophagy under glucose starvation (GS). However, we do not know whether Herp plays any roles in other responses, such as zearalenone (ZEA). In this study, we constructed recombinant lentiviral vectors for Herp shRNA expression and generated stable Herp knockdown RAW 264.7 macrophages. Flow cytometry analysis showed Herp depletion could inhibit cell death induced by ZEA. Western blot analysis revealed that Herp depletion could up-regulate autophagy-related protein LC3-I conversion into LC3-II and the expression of ER stress-related protein CHOP. These results suggest that Herp depletion inhibits cell death by up-regulating autophagy.
Collapse
Affiliation(s)
- Fenglei Chen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Pengfei Lin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Nan Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Diqi Yang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xin Wen
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Dong Zhou
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
34
|
Upadhyay A, Amanullah A, Chhangani D, Mishra R, Mishra A. Selective multifaceted E3 ubiquitin ligases barricade extreme defense: Potential therapeutic targets for neurodegeneration and ageing. Ageing Res Rev 2015; 24:138-59. [PMID: 26247845 DOI: 10.1016/j.arr.2015.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/24/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022]
Abstract
Efficient and regular performance of Ubiquitin Proteasome System and Autophagy continuously eliminate deleterious accumulation of nonnative protiens. In cellular quality control system, E3 ubiquitin ligases are significant employees for defense mechanism against abnormal toxic proteins. Few findings indicate that lack of functions of E3 ubiquitin ligases can be a causative factor of neurodevelopmental disorders, neurodegeneration, cancer and ageing. However, the detailed molecular pathomechanism implying E3 ubiquitin ligases in cellular functions in multifactorial disease conditions are not well understood. This article systematically represents the unique characteristics, molecular nature, and recent developments in the knowledge of neurobiological functions of few crucial E3 ubiquitin ligases. Here, we review recent literature on the roles of E6-AP, HRD1 and ITCH E3 ubiquitin ligases in the neuro-pathobiological mechanisms, with precise focus on the processes of neurodegeneration, and thereby propose new lines of potential targets for therapeutic interventions.
Collapse
|
35
|
Benyair R, Ogen-Shtern N, Lederkremer GZ. Glycan regulation of ER-associated degradation through compartmentalization. Semin Cell Dev Biol 2015; 41:99-109. [DOI: 10.1016/j.semcdb.2014.11.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 12/20/2022]
|
36
|
Ho DV, Chan JY. Induction of Herpud1 expression by ER stress is regulated by Nrf1. FEBS Lett 2015; 589:615-20. [PMID: 25637874 PMCID: PMC10084809 DOI: 10.1016/j.febslet.2015.01.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 12/30/2022]
Abstract
Herpud1 is an ER-localized protein that contributes to endoplasmic reticulum (ER) homeostasis by participating in the ER-associated protein degradation pathway. The Nrf1 transcription factor is important in cellular stress pathways. We show that loss of Nrf1 function results in decreased Herpud1 expression in cells and liver tissues. Expression of Herpud1 increases in response to ER stress, but not in Nrf1 knockout cells. Transactivation studies show that Nrf1 acts through antioxidant response elements located in the Herpud1 promoter, and chromatin immunoprecipitation demonstrates that Herpud1 is a direct Nrf1 target gene. These results indicate that Nrf1 is a transcriptional activator of Herpud1 expression during ER stress, and they suggest Nrf1 is a key player in the regulation of the ER stress response in cells.
Collapse
Affiliation(s)
- Daniel V Ho
- Department of Laboratory Medicine and Pathology, University of California, Irvine, D440 Medical Sciences, Irvine, CA 92697, USA
| | - Jefferson Y Chan
- Department of Laboratory Medicine and Pathology, University of California, Irvine, D440 Medical Sciences, Irvine, CA 92697, USA.
| |
Collapse
|
37
|
Smith MH, Rodriguez EH, Weissman JS. Misfolded proteins induce aggregation of the lectin Yos9. J Biol Chem 2014; 289:25670-7. [PMID: 25086047 DOI: 10.1074/jbc.m114.583344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A substantial fraction of nascent proteins delivered into the endoplasmic reticulum (ER) never reach their native conformations. Eukaryotes use a series of complementary pathways to efficiently recognize and dispose of these terminally misfolded proteins. In this process, collectively termed ER-associated degradation (ERAD), misfolded proteins are retrotranslocated to the cytosol, polyubiquitinated, and degraded by the proteasome. Although there has been great progress in identifying ERAD components, how these factors accurately identify substrates remains poorly understood. The targeting of misfolded glycoproteins in the ER lumen for ERAD requires the lectin Yos9, which recognizes the glycan species found on terminally misfolded proteins. In a role that remains poorly characterized, Yos9 also binds the protein component of ERAD substrates. Here, we identified a 45-kDa domain of Yos9, consisting of residues 22-421, that is proteolytically stable, highly structured, and able to fully support ERAD in vivo. In vitro binding studies show that Yos9(22-421) exhibits sequence-specific recognition of linear peptides from the ERAD substrate, carboxypeptidase Y G255R (CPY*), and binds a model unfolded peptide ΔEspP and protein Δ131Δ in solution. Binding of Yos9 to these substrates results in their cooperative aggregation. Although the physiological consequences of this substrate-induced aggregation remain to be seen, it has the potential to play a role in the regulation of ERAD.
Collapse
Affiliation(s)
- Melanie H Smith
- From the Department of Cellular and Molecular Pharmacology, Graduate Group in Biophysics, California Institute for Quantitative Biosciences, Center for RNA Systems Biology, and Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, California 94158
| | - Edwin H Rodriguez
- From the Department of Cellular and Molecular Pharmacology, Graduate Group in Biophysics, California Institute for Quantitative Biosciences, Center for RNA Systems Biology, and Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, California 94158
| | - Jonathan S Weissman
- From the Department of Cellular and Molecular Pharmacology, Graduate Group in Biophysics, California Institute for Quantitative Biosciences, Center for RNA Systems Biology, and Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, California 94158
| |
Collapse
|
38
|
Yan L, Liu W, Zhang H, Liu C, Shang Y, Ye Y, Zhang X, Li W. Ube2g2-gp78-mediated HERP polyubiquitylation is involved in ER stress recovery. J Cell Sci 2014; 127:1417-27. [PMID: 24496447 DOI: 10.1242/jcs.135293] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A large number of studies have focused on how individual organisms respond to a stress condition, but little attention has been paid to the stress recovery process, such as the endoplasmic reticulum (ER) stress recovery. Homocysteine-induced ER protein (HERP) was originally identified as a chaperone-like protein that is strongly induced upon ER stress. Here we show that, after ER stress induction, HERP is rapidly degraded by Ube2g2-gp78-mediated ubiquitylation and proteasomal degradation. The polyubiquitylation of HERP in vitro depends on a physical interaction between the CUE domain of gp78 and the ubiquitin-like (UBL) domain of HERP, which is essential for HERP degradation in vivo during ER stress recovery. We further show that although HERP promotes cell survival under ER stress, high levels of HERP expression reduce cell viability under oxidative stress conditions, suggesting that HERP plays a dual role in cellular stress adaptation. Together, these results establish the ubiquitin-proteasome-mediated degradation of HERP as a novel mechanism that fine-tunes the stress tolerance capacity of the cell.
Collapse
Affiliation(s)
- Long Yan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Leitman J, Shenkman M, Gofman Y, Shtern NO, Ben-Tal N, Hendershot LM, Lederkremer GZ. Herp coordinates compartmentalization and recruitment of HRD1 and misfolded proteins for ERAD. Mol Biol Cell 2014; 25:1050-60. [PMID: 24478453 PMCID: PMC3967970 DOI: 10.1091/mbc.e13-06-0350] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The unfolded protein response PERK branch induces recruitment of misfolded proteins and the ubiquitin ligase HRD1 to the ER-derived quality control compartment (ERQC), a staging ground for ER-associated degradation (ERAD). This is accomplished by up-regulation of homocysteine-induced ER protein (Herp), which recruits the ERAD complex at the ERQC. A functional unfolded protein response (UPR) is essential for endoplasmic reticulum (ER)-associated degradation (ERAD) of misfolded secretory proteins, reflecting the fact that some level of UPR activation must exist under normal physiological conditions. A coordinator of the UPR and ERAD processes has long been sought. We previously showed that the PKR-like, ER-localized eukaryotic translation initiation factor 2α kinase branch of the UPR is required for the recruitment of misfolded proteins and the ubiquitin ligase HRD1 to the ER-derived quality control compartment (ERQC), a staging ground for ERAD. Here we show that homocysteine-induced ER protein (Herp), a protein highly upregulated by this UPR branch, is responsible for this compartmentalization. Herp localizes to the ERQC, and our results suggest that it recruits HRD1, which targets to ERAD the substrate presented by the OS-9 lectin at the ERQC. Predicted overall structural similarity of Herp to the ubiquitin-proteasome shuttle hHR23, but including a transmembrane hairpin, suggests that Herp may function as a hub for membrane association of ERAD machinery components, a key organizer of the ERAD complex.
Collapse
Affiliation(s)
- Julia Leitman
- Department of Cell Research and Immunology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel Department of Biochemistry and Molecular Biology, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | | | | | | | | | | |
Collapse
|
40
|
Huang CH, Chu YR, Ye Y, Chen X. Role of HERP and a HERP-related protein in HRD1-dependent protein degradation at the endoplasmic reticulum. J Biol Chem 2013; 289:4444-54. [PMID: 24366871 DOI: 10.1074/jbc.m113.519561] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Misfolded proteins of the endoplasmic reticulum (ER) are retrotranslocated to the cytosol and degraded by the proteasome via a process termed ER-associated degradation (ERAD). The precise mechanism of retrotranslocation is unclear. Here, we use several lumenal ERAD substrates targeted for degradation by the ubiquitin ligase HRD1 including SHH (sonic hedgehog) and NHK (null Hong Kong α1-antitrypsin) to study the geometry, organization, and regulation of the HRD1-containing ERAD machinery. We report a new HRD1-associated membrane protein named HERP2, which is homologous to the previously identified HRD1 partner HERP1. Despite sequence homology, HERP2 is constitutively expressed in cells, whereas HERP1 is highly induced by ER stress. We find that these proteins are required for efficient degradation of both glycosylated and nonglycosylated SHH proteins as well as NHK. In cells depleted of HERPs, SHH proteins are largely trapped inside the ER with a fraction of the stabilized SHH protein bound to the HRD1-SEL1L ligase complex. Ubiquitination of SHH is significantly attenuated in the absence of HERPs, suggesting a defect in retrotranslocation. Both HERP proteins interact with HRD1 through a region located in the cytosol. However, unlike its homolog in Saccharomyces cerevisiae, HERPs do not regulate HRD1 stability or oligomerization status. Instead, they help recruit DERL2 to the HRD1-SEL1L complex. Additionally, the UBL domain of HERP1 also seems to have a function independent of DERL2 recruitment in ERAD. Our studies have revealed a critical scaffolding function for mammalian HERP proteins that is required for forming an active retrotranslocation complex containing HRD1, SEL1L, and DERL2.
Collapse
Affiliation(s)
- Chih-Hsiang Huang
- From the Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan 35053, China
| | | | | | | |
Collapse
|
41
|
Bernasconi R, Galli C, Kokame K, Molinari M. Autoadaptive ER-associated degradation defines a preemptive unfolded protein response pathway. Mol Cell 2013; 52:783-93. [PMID: 24239290 DOI: 10.1016/j.molcel.2013.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/23/2013] [Accepted: 10/09/2013] [Indexed: 11/19/2022]
Abstract
Folding-defective proteins must be cleared efficiently from the endoplasmic reticulum (ER) to prevent perturbation of the folding environment and to maintain cellular proteostasis. Misfolded proteins engage dislocation machineries (dislocons) built around E3 ubiquitin ligases that promote their transport across the ER membrane, their polyubiquitylation, and their proteasomal degradation. Here, we report on the intrinsic instability of the HRD1 dislocon and the constitutive, rapid turnover of the scaffold protein HERP. We show that HRD1 dislocon integrity relies on the presence of HRD1 clients that interrupt, in a dose-dependent manner, the UBC6e/RNF5/p97/proteasome-controlled relay that controls HERP turnover. We propose that ER-associated degradation (ERAD) deploys autoadaptive regulatory pathways, collectively defined as ERAD tuning, to rapidly adapt degradation activity to misfolded protein load and to preempt the unfolded protein response (UPR) activation.
Collapse
Affiliation(s)
- Riccardo Bernasconi
- Institute for Research in Biomedicine, Protein Folding and Quality Control, 6500 Bellinzona, Switzerland
| | - Carmela Galli
- Institute for Research in Biomedicine, Protein Folding and Quality Control, 6500 Bellinzona, Switzerland
| | - Koichi Kokame
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center, Osaka 565-8565, Japan
| | - Maurizio Molinari
- Institute for Research in Biomedicine, Protein Folding and Quality Control, 6500 Bellinzona, Switzerland; Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, 1015 Lausanne, Switzerland.
| |
Collapse
|
42
|
Quiroga C, Gatica D, Paredes F, Bravo R, Troncoso R, Pedrozo Z, Rodriguez AE, Toro B, Chiong M, Vicencio JM, Hetz C, Lavandero S. Herp depletion protects from protein aggregation by up-regulating autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3295-3305. [PMID: 24120520 DOI: 10.1016/j.bbamcr.2013.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/21/2013] [Accepted: 09/10/2013] [Indexed: 01/04/2023]
Abstract
Herp is an endoplasmic reticulum (ER) stress inducible protein that participates in the ER-associated protein degradation (ERAD) pathway. However, the contribution of Herp to other protein degradation pathways like autophagy and its connection to other types of stress responses remain unknown. Here we report that Herp regulates autophagy to clear poly-ubiquitin (poly-Ub) protein aggregates. Proteasome inhibition and glucose starvation (GS) led to a high level of poly-Ub protein aggregation that was drastically reduced by stably knocking down Herp (shHerp cells). The enhanced removal of poly-Ub inclusions protected cells from death caused by glucose starvation. Under basal conditions and increasingly after stress, higher LC3-II levels and GFP-LC3 puncta were observed in shHerp cells compared to control cells. Herp knockout cells displayed basal up-regulation of two essential autophagy regulators-Atg5 and Beclin-1, leading to increased autophagic flux. Beclin-1 up-regulation was due to a reduction in Hrd1 dependent proteasomal degradation, and not at transcriptional level. The consequent higher autophagic flux was necessary for the clearance of aggregates and for cell survival. We conclude that Herp operates as a relevant factor in the defense against glucose starvation by modulating autophagy levels. These data may have important implications due to the known up-regulation of Herp in pathological states such as brain and heart ischemia, both conditions associated to acute nutritional stress.
Collapse
Affiliation(s)
- Clara Quiroga
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile; Harvard School of Public Health, Boston, MA, USA
| | - Damian Gatica
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Felipe Paredes
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Roberto Bravo
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Rodrigo Troncoso
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Zully Pedrozo
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Andrea E Rodriguez
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Barbra Toro
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile
| | - Jose Miguel Vicencio
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile; The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Claudio Hetz
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380492, Chile; The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8380492, Chile; Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
43
|
El Khouri E, Le Pavec G, Toledano MB, Delaunay-Moisan A. RNF185 is a novel E3 ligase of endoplasmic reticulum-associated degradation (ERAD) that targets cystic fibrosis transmembrane conductance regulator (CFTR). J Biol Chem 2013; 288:31177-91. [PMID: 24019521 DOI: 10.1074/jbc.m113.470500] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the endoplasmic reticulum (ER), misfolded or improperly assembled proteins are exported to the cytoplasm and degraded by the ubiquitin-proteasome pathway through a process called ER-associated degradation (ERAD). ER-associated E3 ligases, which coordinate substrate recognition, export, and proteasome targeting, are key components of ERAD. Cystic fibrosis transmembrane conductance regulator (CFTR) is one ERAD substrate targeted to co-translational degradation by the E3 ligase RNF5/RMA1. RNF185 is a RING domain-containing polypeptide homologous to RNF5. We show that RNF185 controls the stability of CFTR and of the CFTRΔF508 mutant in a RING- and proteasome-dependent manner but does not control that of other classical ERAD model substrates. Reciprocally, its silencing stabilizes CFTR proteins. Turnover analyses indicate that, as RNF5, RNF185 targets CFTR to co-translational degradation. Importantly, however, simultaneous depletion of RNF5 and RNF185 profoundly blocks CFTRΔF508 degradation not only during translation but also after synthesis is complete. Our data thus identify RNF185 and RNF5 as a novel E3 ligase module that is central to the control of CFTR degradation.
Collapse
Affiliation(s)
- Elma El Khouri
- From the Laboratoire Stress Oxydant et Cancers, Service de Biologie Intégrative et Génétique Moléculaire (SBiGeM), Institut de Biologie et de Technologies de Saclay (IBiTec-S), Commissariat à l'Energie Atomique-Saclay, 91191 Gif-sur-Yvette, Cedex, France
| | | | | | | |
Collapse
|
44
|
Huang CH, Hsiao HT, Chu YR, Ye Y, Chen X. Derlin2 protein facilitates HRD1-mediated retro-translocation of sonic hedgehog at the endoplasmic reticulum. J Biol Chem 2013; 288:25330-25339. [PMID: 23867461 DOI: 10.1074/jbc.m113.455212] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) is an important system that eliminates misfolded proteins from the ER. Three derlins have been implicated in this process, but their precise function remains unknown. In this study, we report that although both derlin1 and derlin2 are capable of binding the ERAD-specific ubiquitin ligase HRD1, they associate with the HRD1-containing complex with different affinities. Accordingly, these derlins have nonredundant functions in ERAD with derlin2 being an essential functional partner for HRD1-mediated ERAD of SHH and NHK. We show that derlin2, but not derlin1 or derlin3, is required for ERAD of both glycosylated and nonglycosylated SHH, as well as NHK. Derlin2 appears to act at a post-targeting step for HRD1-dependent retro-translocation. Without derlin2, the assembly of HRD1 into a functional retro-translocation homo-oligomer proceeds normally, and substrate targeting to the HRD1 complex also occurs. However, the ERAD substrate SHH-C is largely trapped inside the ER lumen. These observations raise the possibility that derlin2 may regulate the movement of substrates through the HRD1-containing retro-translocon. Our study is the first to report that derlin2 functions with HRD1 in ERAD of certain substrates independent of their glycosylation status. The mammalian ERAD system may require multiple derlins that each functions with a distinct E3 partner to eliminate a specific subset of substrates. This is different from the model in Saccharomyces cerevisiae, in which Hrd1p alone is sufficient for retro-translocation.
Collapse
Affiliation(s)
- Chih-Hsiang Huang
- From the Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan 35053, Republic of China
| | - Hui-Ting Hsiao
- From the Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan 35053, Republic of China
| | - Yue-Ru Chu
- From the Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan 35053, Republic of China
| | - Yihong Ye
- the Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Xin Chen
- From the Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan 35053, Republic of China,; the Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan 40402, Republic of China.
| |
Collapse
|
45
|
Wong N, Morahan G, Stathopoulos M, Proietto J, Andrikopoulos S. A novel mechanism regulating insulin secretion involving Herpud1 in mice. Diabetologia 2013; 56:1569-76. [PMID: 23620059 DOI: 10.1007/s00125-013-2908-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 03/18/2013] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes results from beta cell dysfunction after prolonged physiological stress, which causes oversecretion of insulin. We recently found that insulin hypersecretion is mediated by at least two genes. Among mouse models of type 2 diabetes, the DBA/2 mouse strain is more susceptible to diabetes than is the C57BL/6J (B6J) strain. One distinctive feature of the DBA/2 mouse is that it hypersecretes insulin, independent of changes in insulin sensitivity; we identified Nnt as a gene responsible for this trait. METHODS To identify the other gene(s) affecting insulin hypersecretion, we tested a panel of recombinant inbred BXD strains, which have different combinations of B6 and DBA/2 alleles. RESULTS We found that 25% of the BXD strains hypersecreted insulin in response to glucose. Microarray profiling of islets from high- and low-secretor strains showed that at least four genes were differentially expressed. One gene was consistently underexpressed in islets from both DBA/2 and the high-secretor BXD strains. This gene (Herpud1 or Herp) encodes the 54 kDa endoplasmic reticulum stress-inducible protein (HERP) that resides in the integral endoplasmic reticulum membrane. To test directly whether Herpud1 can interact with Nnt, Herpud1 was either knocked down or overexpressed in MIN6 cells. These results showed that when Herpud1 was suppressed, Nnt expression was reduced, while overexpression of Herpud1 led to increased Nnt expression. Furthermore, Herpud1 suppression resulted in significantly decreased glucose-stimulated insulin secretion in the DBA/2 islets but not B6J islets. CONCLUSIONS/INTERPRETATION We conclude that Herpud1 regulates insulin secretion via control of Nnt expression.
Collapse
Affiliation(s)
- N Wong
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Heidelberg Heights, Melbourne, Victoria, 3084, Australia.
| | | | | | | | | |
Collapse
|
46
|
An apicoplast localized ubiquitylation system is required for the import of nuclear-encoded plastid proteins. PLoS Pathog 2013; 9:e1003426. [PMID: 23785288 PMCID: PMC3681736 DOI: 10.1371/journal.ppat.1003426] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 05/01/2013] [Indexed: 01/08/2023] Open
Abstract
Apicomplexan parasites are responsible for numerous important human diseases including toxoplasmosis, cryptosporidiosis, and most importantly malaria. There is a constant need for new antimalarials, and one of most keenly pursued drug targets is an ancient algal endosymbiont, the apicoplast. The apicoplast is essential for parasite survival, and several aspects of its metabolism and maintenance have been validated as targets of anti-parasitic drug treatment. Most apicoplast proteins are nuclear encoded and have to be imported into the organelle. Recently, a protein translocon typically required for endoplasmic reticulum associated protein degradation (ERAD) has been proposed to act in apicoplast protein import. Here, we show ubiquitylation to be a conserved and essential component of this process. We identify apicoplast localized ubiquitin activating, conjugating and ligating enzymes in Toxoplasma gondii and Plasmodium falciparum and observe biochemical activity by in vitro reconstitution. Using conditional gene ablation and complementation analysis we link this activity to apicoplast protein import and parasite survival. Our studies suggest ubiquitylation to be a mechanistic requirement of apicoplast protein import independent to the proteasomal degradation pathway. The apicoplast is an essential parasite organelle derived from an algal endosymbiont. Most apicoplast proteins are nuclear encoded and post-translationally imported. Part of this journey utilizes the endoplasmic reticulum associated degradation or ERAD system of the algal endosymbiont. Typically, the ERAD system is ubiquitylation-dependent and acts in the retrotranslocation across the ER membrane and proteasomal destruction of misfolded secretory proteins. In the apicoplast, this system has been retooled into a protein importer. The apicoplast ERAD system is broadly conserved between most apicomplexans and surprisingly retains the ubiquitylation machine typically associated with destruction. This study brings together biochemical studies in Plasmodium and genetic studies in Toxoplasma. Together they provide significant mechanistic insight into the process of protein import into the apicoplast. We provide evidence that ubiquitylation may be a mechanistic requirement for import and demonstrate it to be essential to the parasite, thus providing new opportunities for drug development.
Collapse
|
47
|
Merulla J, Fasana E, Soldà T, Molinari M. Specificity and Regulation of the Endoplasmic Reticulum-Associated Degradation Machinery. Traffic 2013; 14:767-77. [DOI: 10.1111/tra.12068] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 03/18/2013] [Accepted: 03/23/2013] [Indexed: 02/05/2023]
Affiliation(s)
| | - Elisa Fasana
- Institute for Research in Biomedicine; Protein Folding and Quality Control; CH-6500; Bellinzona; Switzerland
| | - Tatiana Soldà
- Institute for Research in Biomedicine; Protein Folding and Quality Control; CH-6500; Bellinzona; Switzerland
| | | |
Collapse
|
48
|
The role of the unfolded protein response in diabetes mellitus. Semin Immunopathol 2013; 35:333-50. [PMID: 23529219 DOI: 10.1007/s00281-013-0369-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 03/13/2013] [Indexed: 12/13/2022]
Abstract
The endoplasmic reticulum (ER) plays a key role in the synthesis and modification of secretory and membrane proteins in all eukaryotic cells. Under normal conditions, these proteins are correctly folded and assembled in the ER. However, when cells are exposed to environmental factors such as overproduction of ER proteins, viral infections, or glucose deprivation, the secretory and membrane proteins can accumulate in unfolded or misfolded forms in the lumen of the ER, and consequently, cause stress in the ER. To maintain cellular homeostasis, cells induce several responses to ER stress. In mammalian cells, ER stress responses are induced by a diversity of signal pathways. There are three ER-located transmembrane proteins that play important roles in mammalian ER stress responses: activating transcription factor 6, inositol-requiring protein 1, and protein kinase RNA-like endoplasmic reticulum kinase. ER stress is linked to various diseases, including diabetes. This review highlights the particular importance of ER stress-responsive molecules in insulin biosynthesis, glyconeogenesis, insulin resistance, glucose intolerance, and pancreatic β-cell apoptosis. An understanding of the pathogenic mechanism of diabetes from the aspect of ER stress is crucial in formulating therapeutic strategies.
Collapse
|
49
|
Bravo R, Parra V, Gatica D, Rodriguez AE, Torrealba N, Paredes F, Wang ZV, Zorzano A, Hill JA, Jaimovich E, Quest AFG, Lavandero S. Endoplasmic reticulum and the unfolded protein response: dynamics and metabolic integration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:215-90. [PMID: 23317820 DOI: 10.1016/b978-0-12-407704-1.00005-1] [Citation(s) in RCA: 446] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The endoplasmic reticulum (ER) is a dynamic intracellular organelle with multiple functions essential for cellular homeostasis, development, and stress responsiveness. In response to cellular stress, a well-established signaling cascade, the unfolded protein response (UPR), is activated. This intricate mechanism is an important means of re-establishing cellular homeostasis and alleviating the inciting stress. Now, emerging evidence has demonstrated that the UPR influences cellular metabolism through diverse mechanisms, including calcium and lipid transfer, raising the prospect of involvement of these processes in the pathogenesis of disease, including neurodegeneration, cancer, diabetes mellitus and cardiovascular disease. Here, we review the distinct functions of the ER and UPR from a metabolic point of view, highlighting their association with prevalent pathologies.
Collapse
Affiliation(s)
- Roberto Bravo
- Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
de la Monte SM, Re E, Longato L, Tong M. Dysfunctional pro-ceramide, ER stress, and insulin/IGF signaling networks with progression of Alzheimer's disease. J Alzheimers Dis 2012; 30 Suppl 2:S217-29. [PMID: 22297646 DOI: 10.3233/jad-2012-111728] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In Alzheimer's disease (AD), brain insulin and insulin-like growth factor (IGF) resistance and deficiency begin early, and worsen with severity of disease. The factors mediating progression of brain insulin/IGF resistance in AD are not well understood. We hypothesize that AD progression is mediated via negative cross-talk that promotes toxic ceramide generation and endoplasmic reticulum (ER) stress. The rationale is that insulin resistance dysregulates lipid metabolism and promotes ceramide accumulation, and thereby increases inflammation and stress. Consequences include disruption of cytoskeletal function and AβPP-Aβ secretion. The present study correlates AD stage with activation of pro-ceramide genes, ceramide levels, and molecular indices of ER stress in postmortem human brain tissue. The results demonstrated that in AD, brain insulin/IGF resistance was associated with constitutive activation of multiple pro-ceramide genes, increased ceramide levels, and increased expression of pro-ER stress pathway genes and proteins. Expression of several pro-ceramide and pro-apoptotic ER stress pathway molecules increased with AD severity and brain insulin/IGF resistance. In contrast, ER stress molecules that help maintain homeostasis with respect to unfolded protein responses were mainly upregulated in the intermediate rather than late stage of AD. These findings support our hypothesis that in AD, a triangulated mal-signaling network initiated by brain insulin/IGF resistance is propagated by the dysregulation of ceramide and ER stress homeostasis, which themselves promote insulin resistance. Therefore, once established, this reverberating loop must be targeted using multi-pronged approaches to disrupt the AD neurodegeneration cascade.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology (Neuropathology), Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA. Suzanne DeLaMonte
| | | | | | | |
Collapse
|